Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 10(1): 80-85, 2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30655951

ABSTRACT

Potent covalent inhibitors of Bruton's tyrosine kinase (BTK) based on an aminopyrazole carboxamide scaffold have been identified. Compared to acrylamide-based covalent reactive groups leading to irreversible protein adducts, cyanamide-based reversible-covalent inhibitors provided the highest combined BTK potency and EGFR selectivity. The cyanamide covalent mechanism with BTK was confirmed through enzyme kinetic, NMR, MS, and X-ray crystallographic studies. The lead cyanamide-based inhibitors demonstrated excellent kinome selectivity and rat pharmacokinetic properties.

2.
Biochem J ; 460(2): 211-22, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24593284

ABSTRACT

ITK (interleukin-2-inducible T-cell kinase) is a critical component of signal transduction in T-cells and has a well-validated role in their proliferation, cytokine release and chemotaxis. ITK is an attractive target for the treatment of T-cell-mediated inflammatory diseases. In the present study we describe the discovery of kinase inhibitors that preferentially bind to an allosteric pocket of ITK. The novel ITK allosteric site was characterized by NMR, surface plasmon resonance, isothermal titration calorimetry, enzymology and X-ray crystallography. Initial screening hits bound to both the allosteric pocket and the ATP site. Successful lead optimization was achieved by improving the contribution of the allosteric component to the overall inhibition. NMR competition experiments demonstrated that the dual-site binders showed higher affinity for the allosteric site compared with the ATP site. Moreover, an optimized inhibitor displayed non-competitive inhibition with respect to ATP as shown by steady-state enzyme kinetics. The activity of the isolated kinase domain and auto-activation of the full-length enzyme were inhibited with similar potency. However, inhibition of the activated full-length enzyme was weaker, presumably because the allosteric site is altered when ITK becomes activated. An optimized lead showed exquisite kinome selectivity and is efficacious in human whole blood and proximal cell-based assays.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Adenosine Triphosphate/pharmacology , Allosteric Regulation , Allosteric Site , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation/drug effects , Protein Structure, Tertiary , Surface Plasmon Resonance
3.
Bioorg Med Chem Lett ; 23(3): 767-72, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23265881

ABSTRACT

Identification of a series of imidazo[4,5-c]pyridin-4-one derivatives that act as dual angiotensin II type 1 (AT1) receptor antagonists and peroxisome proliferator-activated receptor-γ (PPARγ) partial agonists is described. Starting from a known AT1 antagonist template, conformational restriction was introduced by incorporation of an indane ring that when combined with appropriate substitution at the imidazo[4,5-c]pyridin-4-one provided novel series 5 possessing the desired dual activity. The mode of interaction of this series with PPARγ was corroborated through the X-ray crystal structure of 12b bound to the human PPARγ ligand binding domain. Modulation of activity at both receptors through substitution at the pyridone nitrogen led to the identification of potent dual AT1 antagonists/PPARγ partial agonists. Among them, 21b was identified possessing potent dual pharmacology (AT1 IC(50) = 7 nM; PPARγ EC(50) = 295 nM, 27% max) and good ADME properties.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/chemical synthesis , Angiotensin II Type 1 Receptor Blockers/pharmacology , PPAR gamma/metabolism , Pyridines/chemical synthesis , Pyridines/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II Type 1 Receptor Blockers/chemistry , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzoates/chemistry , Benzoates/pharmacology , Crystallography, X-Ray , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , PPAR gamma/agonists , Protein Binding/drug effects , Pyridines/chemistry , Pyridones/chemical synthesis , Pyridones/chemistry , Pyridones/pharmacology , Telmisartan
4.
J Med Chem ; 54(12): 4219-33, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21557540

ABSTRACT

Mining of an in-house collection of angiotensin II type 1 receptor antagonists to identify compounds with activity at the peroxisome proliferator-activated receptor-γ (PPARγ) revealed a new series of imidazo[4,5-b]pyridines 2 possessing activity at these two receptors. Early availability of the crystal structure of the lead compound 2a bound to the ligand binding domain of human PPARγ confirmed the mode of interaction of this scaffold to the nuclear receptor and assisted in the optimization of PPARγ activity. Among the new compounds, (S)-3-(5-(2-(1H-tetrazol-5-yl)phenyl)-2,3-dihydro-1H-inden-1-yl)-2-ethyl-5-isobutyl-7-methyl-3H-imidazo[4,5-b]pyridine (2l) was identified as a potent angiotensin II type I receptor blocker (IC(50) = 1.6 nM) with partial PPARγ agonism (EC(50) = 212 nM, 31% max) and oral bioavailability in rat. The dual pharmacology of 2l was demonstrated in animal models of hypertension (SHR) and insulin resistance (ZDF rat). In the SHR, 2l was highly efficacious in lowering blood pressure, while robust lowering of glucose and triglycerides was observed in the male ZDF rat.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/chemical synthesis , Antihypertensive Agents/chemical synthesis , Hypoglycemic Agents/chemical synthesis , Imidazoles/chemical synthesis , PPAR gamma/agonists , Pyridines/chemical synthesis , Administration, Oral , Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Biological Availability , Blood Glucose/analysis , Crystallography, X-Ray , Drug Partial Agonism , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Imidazoles/chemistry , Imidazoles/pharmacology , Insulin Resistance , Male , Models, Molecular , Pyridines/chemistry , Pyridines/pharmacology , Radioligand Assay , Rats , Rats, Inbred SHR , Stereoisomerism , Structure-Activity Relationship , Transcriptional Activation , Triglycerides/blood
5.
Proc Natl Acad Sci U S A ; 107(34): 15240-5, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20696890

ABSTRACT

Circadian pacemaking requires the orderly synthesis, posttranslational modification, and degradation of clock proteins. In mammals, mutations in casein kinase 1 (CK1) epsilon or delta can alter the circadian period, but the particular functions of the WT isoforms within the pacemaker remain unclear. We selectively targeted WT CK1epsilon and CK1delta using pharmacological inhibitors (PF-4800567 and PF-670462, respectively) alongside genetic knockout and knockdown to reveal that CK1 activity is essential to molecular pacemaking. Moreover, CK1delta is the principal regulator of the clock period: pharmacological inhibition of CK1delta, but not CK1epsilon, significantly lengthened circadian rhythms in locomotor activity in vivo and molecular oscillations in the suprachiasmatic nucleus (SCN) and peripheral tissue slices in vitro. Period lengthening mediated by CK1delta inhibition was accompanied by nuclear retention of PER2 protein both in vitro and in vivo. Furthermore, phase mapping of the molecular clockwork in vitro showed that PF-670462 treatment lengthened the period in a phase-specific manner, selectively extending the duration of PER2-mediated transcriptional feedback. These findings suggested that CK1delta inhibition might be effective in increasing the amplitude and synchronization of disrupted circadian oscillators. This was tested using arrhythmic SCN slices derived from Vipr2(-/-) mice, in which PF-670462 treatment transiently restored robust circadian rhythms of PER2::Luc bioluminescence. Moreover, in mice rendered behaviorally arrhythmic by the Vipr2(-/-) mutation or by constant light, daily treatment with PF-670462 elicited robust 24-h activity cycles that persisted throughout treatment. Accordingly, selective pharmacological targeting of the endogenous circadian regulator CK1delta offers an avenue for therapeutic modulation of perturbed circadian behavior.


Subject(s)
Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Circadian Rhythm/physiology , Animals , Base Sequence , Casein Kinase 1 epsilon/physiology , Casein Kinase Idelta/deficiency , Casein Kinase Idelta/genetics , Casein Kinase Idelta/physiology , Circadian Rhythm/drug effects , Gene Knockdown Techniques , In Vitro Techniques , Mice , Mice, Knockout , Mice, Transgenic , Period Circadian Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/deficiency , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/physiology
6.
Bioorg Med Chem ; 17(20): 7113-25, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19783444

ABSTRACT

The synthesis of a new series of phenylpropanoic acid derivatives incorporating an heteroaryl group at the alpha-position and their evaluation for binding and activation of PPARalpha and PPARgamma are presented in this report. Among the new compounds, (S)-3-{4-[3-(5-methyl-2-phenyl-oxazol-4-yl)-propyl]-phenyl}-2-1,2,3-triazol-2-yl-propionic acid (17j), was identified as a potent human PPARalpha/gamma dual agonist (EC(50)=0.013 and 0.061 microM, respectively) with demonstrated oral bioavailability in rat and dog. 17j was shown to decrease insulin levels, plasma glucose, and triglycerides in the ZDF female rat model. In the human apolipoprotein A-1/CETP transgenic mouse model 17j produced increases in hApoA1 and HDL-C and decreases in plasma triglycerides. The increased potency for binding and activation of both PPAR subtypes observed with 17j when compared to previous analogs in this series was explained based on results derived from crystallographic and modeling studies.


Subject(s)
PPAR alpha/agonists , PPAR gamma/agonists , Propionates/chemical synthesis , Propionates/pharmacology , Animals , Biological Availability , Blood Glucose/analysis , Crystallography, X-Ray , Dogs , Drug Evaluation, Preclinical , Female , Insulin/blood , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Mice, Transgenic , Propionates/pharmacokinetics , Rats , Triglycerides/blood
7.
Bioorg Med Chem ; 16(9): 4883-907, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18394907

ABSTRACT

A new series of alpha-aryl or alpha-heteroarylphenyl propanoic acid derivatives was synthesized that incorporate acetylene-, ethylene-, propyl-, or nitrogen-derived linkers as a replacement of the commonly used ether moiety that joins the central phenyl ring with the lipophilic tail. The effect of these modifications in the binding and activation of PPARalpha and PPARgamma was first evaluated in vitro. Compounds possessing suitable profiles were then evaluated in the ob/ob mouse model of type 2 diabetes. The propylene derivative 40 and the propyl derivative 53 demonstrated robust plasma glucose lowering activity in this model. Compound 53 was also evaluated in male Zucker diabetic fatty rats and was found to achieve normalization of glucose, triglycerides, and insulin levels. An X-ray crystal structure of the complex of 53 with the PPARgamma-ligand-binding domain was obtained and discussed in this report.


Subject(s)
PPAR alpha/agonists , PPAR gamma/agonists , Phenylpropionates , Administration, Oral , Animals , Crystallography, X-Ray , Disease Models, Animal , Male , Mice , Mice, Obese , Models, Molecular , Molecular Structure , Phenylpropionates/chemical synthesis , Phenylpropionates/chemistry , Phenylpropionates/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Zucker , Stereoisomerism , Structure-Activity Relationship
8.
Acta Crystallogr D Biol Crystallogr ; 64(Pt 3): 321-30, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18323627

ABSTRACT

Aspartate-beta-semialdehyde dehydrogenase (ASADH) is an essential enzyme that is found in bacteria, fungi and plants but not in humans. ASADH produces the first branch-point metabolite in the biosynthetic pathways that lead to the production of lysine, threonine, methionine and isoleucine as well as the cell-wall precursor diaminopimelate. As a consequence, ASADH appears to be an excellent target for the development of novel antibiotics, especially for Gram-negative bacteria that require diaminopimelate for cell-wall biosynthesis. In contrast to the Gram-negative ASADHs, which readily formed well diffracting crystals, the second isoform of aspartate-beta-semialdehyde dehydrogenase from Vibrio cholerae (vcASADH2) was less well behaved in initial crystallization trials. In order to obtain good-quality single crystals of vcASADH2, a buffer-optimization protocol was used in which the initial purification buffer was exchanged into a new condition derived from a pre-crystalline hit. The unliganded structure of vcASADH2 has been determined to 2.2 A resolution to provide additional insight into the structural and functional evolution of the ASADH enzyme family. The overall fold and domain organization of this new structure is similar to the Gram-negative, Gram-positive and archeal ASADH structures determined previously, despite having less than 50% sequence identity to any of these family members. The substrate-complex structure reveals that the binding of L-aspartate-beta-semialdehyde (ASA) to vcASADH2 is accommodated by structural changes in the amino-acid binding site and in the helical subdomain that is involved in the dimer interface. Structural alignments show that this second isoform from Gram-negative V. cholerae most closely resembles the ASADH from a Gram-positive organism and is likely to bind the coenzyme in a different conformation to that observed in the other V. cholerae isoform.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/chemistry , Protein Conformation , Vibrio cholerae/enzymology , Crystallography, X-Ray , Models, Molecular , Protein Structure, Tertiary , Substrate Specificity
9.
Protein Sci ; 16(12): 2657-66, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18029420

ABSTRACT

N-Acetylglucosamine-1-phosphate uridyltransferase (GlmU) catalyzes the first step in peptidoglycan biosynthesis in both Gram-positive and Gram-negative bacteria. The products of the GlmU reaction are essential for bacterial survival, making this enzyme an attractive target for antibiotic drug discovery. A series of Haemophilus influenzae GlmU (hiGlmU) structures were determined by X-ray crystallography in order to provide structural and functional insights into GlmU activity and inhibition. The information derived from these structures was combined with biochemical characterization of the K25A, Q76A, D105A, Y103A, V223A, and E224A hiGlmU mutants in order to map these active-site residues to catalytic activity of the enzyme and refine the mechanistic model of the GlmU uridyltransferase reaction. These studies suggest that GlmU activity follows a sequential substrate-binding order that begins with UTP binding noncovalently to the GlmU enzyme. The uridyltransferase active site then remains in an open apo-like conformation until N-acetylglucosamine-1-phosphate (GlcNAc-1-P) binds and induces a conformational change at the GlcNAc-binding subsite. Following the binding of GlcNAc-1-P to the UTP-charged uridyltransferase active site, the non-esterified oxygen of GlcNAc-1-P performs a nucleophilic attack on the alpha-phosphate group of UTP. The new data strongly suggest that the mechanism of phosphotransfer in the uridyltransferase reaction in GlmU is primarily through an associative mechanism with a pentavalent phosphate intermediate and an inversion of stereochemistry. Finally, the structural and biochemical characterization of the uridyltransferase active site and catalytic mechanism described herein provides a basis for the structure-guided design of novel antibacterial agents targeting GlmU activity.


Subject(s)
Haemophilus influenzae/enzymology , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Binding Sites , Catalysis , Crystallography, X-Ray , Ligands , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Conformation , Protein Structure, Tertiary , Uridine/chemistry , Uridine/metabolism , Uridine Triphosphate/metabolism
10.
J Med Chem ; 50(21): 5090-102, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17880056

ABSTRACT

A new series of MEK1 inhibitors, the 4-anilino-5-carboxamido-2-pyridones, were designed and synthesized using a combination of medicinal chemistry, computational chemistry, and structural elucidation. The effect of variation in the carboxamide side chain, substitution on the pyridone nitrogen, and replacement of the 4'-iodide were all investigated. This study afforded several compounds which were either equipotent or more potent than the clinical candidate CI-1040 (1) in an isolated enzyme assay, as well as murine colon carcinoma (C26) cells, as measured by suppression of phosphorylated ERK substrate. Most notably, pyridone 27 was found to be more potent than 1 in vitro and produced a 100% response rate at a lower dose than 1, when tested for in vivo efficacy in animals bearing C26 tumors.


Subject(s)
Amides/chemical synthesis , Aniline Compounds/chemical synthesis , Antineoplastic Agents/chemical synthesis , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Pyridones/chemical synthesis , Amides/chemistry , Amides/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 2/chemistry , Male , Mice , Models, Molecular , Neoplasm Transplantation , Phosphorylation , Pyridones/chemistry , Pyridones/pharmacology , Rats , Structure-Activity Relationship
12.
Acta Crystallogr D Biol Crystallogr ; 63(Pt 3): 381-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17327675

ABSTRACT

The structure of a transition-state analog complex of a highly soluble mutant (R134K) of rabbit muscle creatine kinase (rmCK) has been determined to 1.65 A resolution in order to elucidate the structural changes that are required to support and regulate catalysis. Significant structural asymmetry is seen within the functional homodimer of rmCK, with one monomer found in a closed conformation with the active site occupied by the transition-state analog components creatine, MgADP and nitrate. The other monomer has the two loops that control access to the active site in an open conformation and only MgADP is bound. The N-terminal region of each monomer makes a substantial contribution to the dimer interface; however, the conformation of this region is dramatically different in each subunit. Based on this structural evidence, two mutational modifications of rmCK were conducted in order to better understand the role of the amino-terminus in controlling creatine kinase activity. The deletion of the first 15 residues of rmCK and a single point mutant (P20G) both disrupt subunit cohesion, causing the dissociation of the functional homodimer into monomers with reduced catalytic activity. This study provides support for a structural role for the amino-terminus in subunit association and a mechanistic role in active-site communication and catalytic regulation.


Subject(s)
Creatine Kinase, MM Form/chemistry , Amino Acid Sequence , Animals , Binding Sites , Catalysis , Creatine Kinase, MM Form/genetics , Crystallography, X-Ray , Dimerization , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Protein Structure, Tertiary , Rabbits , Sequence Alignment
14.
J Mol Biol ; 353(5): 1055-68, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16225889

ABSTRACT

The structure of aspartate-beta-semialdehyde dehydrogenase (ASADH) from Methanococcus jannaschii has been determined to 2.3 angstroms resolution using multiwavelength anomalous diffraction (MAD) phasing of a selenomethionine-substituted derivative to define a new branch in the family of ASADHs. This new structure has a similar overall fold and domain organization despite less than 10% conserved sequence identity with the bacterial enzymes. However, the entire repertoire of functionally important active site amino acid residues is conserved, suggesting an identical catalytic mechanism but with lower catalytic efficiency. A new coenzyme-binding conformation and dual NAD/NADP coenzyme specificity further distinguish this archaeal branch from the bacterial ASADHs. Several structural differences are proposed to account for the dramatically enhanced thermostability of this archaeal enzyme. Finally, the intersubunit communication channel connecting the active sites in the bacterial enzyme dimer has been disrupted in the archaeal ASADHs by amino acid changes that likely prevent the alternating sites reactivity previously proposed for the bacterial ASADHs.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/chemistry , Methanococcus/enzymology , Archaeal Proteins/chemistry , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Enzyme Stability , Models, Molecular , Protein Structure, Tertiary , Protein Subunits , Selenomethionine , Sequence Alignment
15.
Nat Struct Mol Biol ; 11(12): 1192-7, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15543157

ABSTRACT

MEK1 and MEK2 are closely related, dual-specificity tyrosine/threonine protein kinases found in the Ras/Raf/MEK/ERK mitogen-activated protein kinase (MAPK) signaling pathway. Approximately 30% of all human cancers have a constitutively activated MAPK pathway, and constitutive activation of MEK1 results in cellular transformation. Here we present the X-ray structures of human MEK1 and MEK2, each determined as a ternary complex with MgATP and an inhibitor to a resolution of 2.4 A and 3.2 A, respectively. The structures reveal that MEK1 and MEK2 each have a unique inhibitor-binding pocket adjacent to the MgATP-binding site. The presence of the potent inhibitor induces several conformational changes in the unphosphorylated MEK1 and MEK2 enzymes that lock them into a closed but catalytically inactive species. Thus, the structures reported here reveal a novel, noncompetitive mechanism for protein kinase inhibition.


Subject(s)
Enzyme Inhibitors/pharmacology , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/chemistry , MAP Kinase Kinase 2/metabolism , Binding Sites , Conserved Sequence , Dimerization , Enzyme Inhibitors/chemistry , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Models, Molecular , Molecular Structure , Protein Structure, Quaternary , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...